CA2185599C - Methods for the manufacture of terbinafine, trans-n-methyl-n-(1-naphthylmethyl)-6,6-dimethylhept-2-en-4 -ynyl-1-amine and pharmaceutically acceptable salt thereof, intermediates useful in the manufacture thereof, novel n-methyl-n-(1-naphthylmethyl)-6,6-dimethyl-2-hydroxyheptan- 4-ynyl-1-amine as potential antimycotic agents - Google Patents

Methods for the manufacture of terbinafine, trans-n-methyl-n-(1-naphthylmethyl)-6,6-dimethylhept-2-en-4 -ynyl-1-amine and pharmaceutically acceptable salt thereof, intermediates useful in the manufacture thereof, novel n-methyl-n-(1-naphthylmethyl)-6,6-dimethyl-2-hydroxyheptan- 4-ynyl-1-amine as potential antimycotic agents

Info

Publication number
CA2185599C
CA2185599C CA002185599A CA2185599A CA2185599C CA 2185599 C CA2185599 C CA 2185599C CA 002185599 A CA002185599 A CA 002185599A CA 2185599 A CA2185599 A CA 2185599A CA 2185599 C CA2185599 C CA 2185599C
Authority
CA
Canada
Prior art keywords
methyl
naphthylmethyl
ynyl
amine
dimethyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
CA002185599A
Other languages
French (fr)
Other versions
CA2185599A1 (en
Inventor
Khashayar Karimian
Regis Chung Soon Hin Leung-Toung
Yiwei Li
Fat Tim Tam
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Apotex Inc
Original Assignee
Apotex Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Apotex Inc filed Critical Apotex Inc
Publication of CA2185599A1 publication Critical patent/CA2185599A1/en
Application granted granted Critical
Publication of CA2185599C publication Critical patent/CA2185599C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C213/00Preparation of compounds containing amino and hydroxy, amino and etherified hydroxy or amino and esterified hydroxy groups bound to the same carbon skeleton
    • C07C213/08Preparation of compounds containing amino and hydroxy, amino and etherified hydroxy or amino and esterified hydroxy groups bound to the same carbon skeleton by reactions not involving the formation of amino groups, hydroxy groups or etherified or esterified hydroxy groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C209/00Preparation of compounds containing amino groups bound to a carbon skeleton
    • C07C209/68Preparation of compounds containing amino groups bound to a carbon skeleton from amines, by reactions not involving amino groups, e.g. reduction of unsaturated amines, aromatisation, or substitution of the carbon skeleton

Abstract

Process for the manufacture of naphthylmethylamine derivatives and amongst them terbinafme as potential antimycotic agents.

Description

METHODS FOR THE MANUFACTURE OF TERBINAFINE, TRANS-N-METHYL-N-(1-NAPHTHYLMETHYL)-6,6-DIMETHYLHEPT-2-EN-4-THEREOF. INTERMEDIATES USEFUL IN THE MANUFACTURE
THEREOF, NOVEL N-METHYL-N-(1-NAPHTHYLMETHYL)-6,6-ANTIMYCOTIC AGENTS.
FIELD OF THE INVENTION
This invention relates to novel processes for the manufacture of traps-N-methyl-N-( 1-naphthylmethyl)-6,6-dimethylhept-2-en-4-ynyl-1-amine, novel intermediates of formula II useful in the manufacture of such naphthylmethylamines, and novel processes for the manufacture of the intermediates used.
R~ OH R2 N
/ I \
wherein: ~ ~ 1 ) R' is lower alkyl R2 is alkyl, branched alkyl , aryl.
Alkyl groups include straight and branched chain hydrocarbon radicals having 1 to 8 carbon atoms.
The lower/alkyl groups include straight and branched chain hydrocarbon radicals from 1 to 4 carbon atoms.
Aryl as used herein include phenyl or naphthyl.
According to further aspects of this invention, there are provided methods for the conversion of compounds of formula II to terbinafine and naphthylmethylamines derivatives of formula I.
R~ R2 N ~~~
/ \
\ /
wherein:
R' is lower alkyl RZ is alkyl, branched alkyl, aryl.
A third aspect of this invention relates to a process of reacting an aldehyde of formula (V) with a Wittig reagent of formula (VI), or with a reagent of formula (VII) to give compound of formula (I), RI
N ECHO

/ \ Br- + / R (Et0)2P
Ph3P
(VII) (V) (VI) wherein:
R' and R2 have the same definition as described above.
A fourth aspect of this invention concerns the use of compounds of formula (IV) as a precursor in the synthesis of compounds of formula (I), wherein R' has the same definition as above and R3, R4 are independently lower alkyl, or when R3 and R4 are taken together, form an ethylene linkage of -CHZCHZ-.
R~ OR3 i N~OR4 / I \
(IV) BACKGROUND OF INVENTION
This invention relates to certain N-alkyl-N-(1-naphthylmethyl)-2-hydroxyalkyl-4-ynyl-1-amines as potential antimycotic agents. Several articles have been published emphasising the pharmaceutical properties of terbinafine, see Petranyl, G. et. al., Science, 1984, 24, 1239; Stutz.A. et. al. , J. Med.
Chem., 1984, 27, 1539. Terbinafme is a powerful inhibitor of fungal squalene epoxidase which serves as a key enzyme in fungal steroid biosynthesis. Terbinafine is an approved topical antifungal agent. It has been prepared by three different methods.
In the first method, described in Canadian Patent 1 157 023, N-alkylation of N-methyl-N-naphthylamine with traps-6,6-dimethylhept-2-en-4-ynyl-1-bromide results in the formation of terbinafine. The traps-6,6-dimethylhept-2-en-4-ynyl-1-bromide is prepared from 6,6-dimethylhepten-4-ynyl-3-of and hydrogen bromide as illustrated in Scheme 1.

1 ) BuLi 2) acrolein CH Br~

I
NH
O O N~
Terbinafine (I) Scheme 1 In the second method also described in Canadian Patent 1 157 023, N-methyl-N-(1-naphthylmethyl)-6,6-dimethylhept-2-4-diynyl-1-amine is reduced by catalytic hydrogenation to give terbinafine. The key intermediate N-methyl-N-(1-naphthylmethyl)-6,6-dimethylhept-2-4-diynyl-1-amine is prepared from N-methyl-N-naphthylamine and 6,6-dimethylhept-2-4-diyne, or from the acetylene coupling reaction between N-methyl-N-naphthylmethylpropargyl amine and tert-butylacetylene bromide as depicted in Scheme 2.
I
NH
CuCI, (CHzO)n I
OO O N
N
CuCI
w Br N 00 Terbinafine (I) Scheme 2 Canadian Patent 1 157 023, further relates to a 3rd method of preparing terbinafine by reductive amination of the naphthylamine with traps-6,6-dimethylhept-2-en-4-yn-1-al in the presence of formaldehyde and sodium borohydride. The reaction is described in Scheme 3.

1 ) NaBH4 // \ _~ N
I \ 2) HCHO
H NaBH4 Scheme 3 When compared to the above processes, the applicant's invention introduces a number of advantages over the existing processes:
First, it affords terbinafine in considerably higher yields than existing procedures.
Second, it is amenable to industrial scale production since terbinafine can be made in three steps from commercially available starting material.
Third, it avoids the use of intermediates such as traps-6,6-dimethylhept-2-en-4-yn-1-al, 6,6-dimethylhept-2-4-diyne, and traps-6,6-dimethylhept-2-en-4-ynyl-1-bromide. All of those intermediates derives from multistep synthesis thereby rending the process more expensive.
Fourth, it avoids the use of acreloin (Scheme 1 ) which is a very hazardous chemical and not commercially available in pure form.
Fifth, it avoids the use of toxic and volatile reagents such as formaldehyde.
Therefore, one object of the present invention is to provide novel process for the production of terbinafine from readily available, inexpensive and relatively safe starting materials. Other objects of this invention can be recognized by those skill in the art from the summary of invention and detailed description of embodiments thereof.
SUMMARY OF INVENTION
According to one aspect of the present invention, a process is provided to make terbinafine which comprises of the steps of conversion of N-alkyl-N-naphthylmethylamine to their corresponding 2,3-epoxypropane (III) as shown in Scheme 4. Compound III is then reacted with lithium alkylacetylide in the presence of lewis acid to give N-alkyl-N-( 1-naphthylmethyl)-2-hydroxyheptan-4-ynyl-1-amine, a compound of formula II. Dehydration of the alcohol II affords a mixture of E and Z-terbinafine.

I RI
R
N O
NH.HCI
O~CI /Et3N/THF E
a BF3.OEt2/THF
~itt>
Ri RI
OH
I ) MsC I/Et; N
2) DBU/tol/A
TERBINAFINE
(II) Scheme 4 ~ I ) A second process includes the reaction between an aldehyde of formula (V) with a Wittig reagent (VI), or with a reagent (VII) to afford the enyne 15 compound of formula I (Scheme 5). Reaction ofN-alkyl-N-naphthylmethylamine with bromo acetaldehyde dialkyl acetal gives the compound of formula (IV), which can be hydrolysed in acid to give the aldehyde (V) I

NH y N O Ra 20 / \ OR3 / + Br~OR4 -' \ / (IV) RI
I
NACHO RI
i /
25 ~_ / \ Br- R2 N~ /
/ Ph3P+ ~ / \
(vI) \ I /
(v) n-BuLi/THF ( 1 ) or NH3/CH;CN

_g_ DETAILED DESCRIPTION OF INVENTION
In the first process of this invention, N-methyl-N-naphthylmethylamine is reacted with epichlorohydrin in the presence of a strong base such as sodium hydroxide, sodium methoxide or tetrabutylammonium hydroxide in alcohol to S give the N-methyl-N-naphthylmethyl-2,3-epoxypropane (III). N-Methyl-N-naphthylmethylamine could, in principle, react with epichlorohydrin to give the diamine derivative 1,3-di-(N-methyl-N-naphthylmethylamino)propan-2-of because the epoxide (III) may undergo further reaction with excess amine. To prevent the formation of this undesirable side product, N-methyl-N-naphthylmethyl is reacted with excess epichlorohydrin to give the N-methyl-N-naphthylmethyl-2,3-epoxypropane (III) as the major product. The excess epichlorohydrin can be removed by distillation. The preferred condition requires the use of 5 to 10 equivalents of epichlorohydrin, in the presence of sodium hydroxide, in methanol at 60°C for a period of 3 hrs. The epoxide (III) is isolated 1 S by conventional means.
The epoxide (III) does not react with lithium tert-butylacetylene in an inert solvent such as tetrahydrofuran. However, the reaction proceeds smoothly in presence of boron trifluoride ethereate at -78°C. The most preferred condition for this transformation requires the mixing of lithum tert-butylacetylene and epoxide (III) at -20°C, followed by the addition of boron triflouride ethereate. The product can be isolated by conventional methods.
The hydroxyl function of the resulting alcohol (II) is converted to the corresponding methanesulfonate or tosylate, or other good leaving groups, in the presence of a base such as triethylamine, and methanesulfonyl chloride or toluenesulfonyl chloride at low temperature, preferrably at 0°C.
Treatment of the resulting material with a strong base such as 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU) or 1.~-diazabicvclo[4.3.0]undec-7-ene (DBN), tort-butoxide. potassium hydroxide and the like affords the E/Z terbinaline as a mixture. The E isomer is separated by recrystallization as a hydrochloride salt. In another embodiment.
the alcohol (II) can be directly dehydrated to give a mixture of E/Z terbinafine by heating in dimethyl sulfoxide (DMSO), preferrably at reflux temperature. In a further embodiment, the alcohol (II) can be dehydrated under acidic conditions to afford a mixture of E/Z terbinafine. For example, heating the alcohol (II) in DMSO containing silica gel and p-toluenesulfonic acid or heating the alcohol (II) in toluene containing Amberlyst* 15.
-In the second process ofthis invention, N-methyl-N-naphthylmethylamine reacts with bromoacetaldehyde dialkylacetal in the presence of a base to give a compound of formula (IV) which undergoes acid hydrolysis to afford the aldehyde of formula (V). Examples of bases used in the N-alkylation are sodium hydroxide and potassium carbonate. Compound of formula (IV) will undergo acid hydrolysis with diluted mineral acid such as dilute hydrochloric acid at elevated temperature to give the aldehyde. Alternatively, the hemiacetal (IV) can be deprotected by stirring the compound in acetone in the presence of p-toluenesulfonic acid at room temperature. The aldehyde reacts with the Wittig reagent (VI) or the reagent (VII) in the presence of base such as ammonia in acetonitrile or n-buyllithium in tetrahydrofuran to give the E/Z terbinafine as a mixture.
The starting materials are prepared following processes well documented in the art. The Wittig reagent (VI) is prepared from 1-bromoalkyne and triphenylphosphine according to literature procedures, see Eiter, K.; Oediger, H.., Liebigs Ann. Chem. 1965, 62, 682, Corey, E.J.; Ruden, R.A., Tetrahedron Lett. 1973. 1495 and 1-bromoalkyne can be prepared by the procedures outlined * trademark in Brandsma, L. et. al, in Synthesis of Acetylenes, Allenes and Cumulenes, a laboratory manual, p. 221, Elsevier Scientific Publishing Company, 1981.
Preparation of compound (IV) is carried out according to the procedure reported in Sandier, S.R. and Karo, W., in Organic Functional Group Preparation, p.
385, Academic Press, Inc., 1983 and the hydrolysis of compound (IV) to aldehyde (V) is accomplished following standard procedures taken from Greene, T.W., in Protective Groups in Organic Synthesis, ppl 19-127, John Wiley &
Sons, 1981.
The present invention will be more fully understood by the following examples which illustrate the invention, but are not considered limiting to the scope of the invention.
Example I
N-Meth, 1-~phthylmeth,~,3-epoxy~ropane (III
(i) To an ice-cooled solution of N-methyl-1-naphthalenemethylamine hydrochloride (2.1 g) in methanol (40 mL) and water ( 10 mL) was added sodium hydroxide powder (2 g) followed by dropwise addition of epichlorohydrin (8 mL). The mixture was heated at 60°C for 3 h then cooled to room temperature.
Volatile materials were removed in vacuo and the residue was taken up in ethyl acetate and washed with water. The organic phase was collected, dried over sodium sulfate, filtered and evaporated to dryness. The crude mixture was purified by flash chromatrography on silica gel (grade 9385, Merck, 230-400 mesh, 60 A) using a solvent gradient of a mixture of hexane and ethyl acetate (95:5, 90:10 and 85:15) as eluent, thereby affording the title compound (III) ( 1.85 g, 81.5%) as an oil.
'H-NMR (CDC13) 8 (ppm): 8.37 (m, 1 H); 7.80-7.91 (m, 2H); 7.44-7.58 (m, 4H);
4.11 (d, J=13.0 Hz, 1 H); 3.92 (d, J=13 Hz, 1 H); 3.17 (m, 1 H); 2.85 (dd, J=13.4 and 3.6 Hz, 1 H); 2.78 (m, 1 H); 2.49 (dd, J=5.0 and 2.7 Hz, 1 H); 2.44 (dd, J=13 .4 and 6.5 Hz) and 2.40 (s, 3H).
'3C-NMR (CDCl3) 8 (ppm): 134.6, 133.9, 132.5, 128.5, 128.1, 127.5, 125.9, 125.7, 125.1, 124.7, 61.0, 60.0, 50.9, 45.1, 43.2.
HRMS: calc. for C,SH,~NO 227.1310 found 227.1315.
(ii) Similarly, the title compound (III) can be obtained when sodium methoxide is employed as base.
(iii) Similarly, the title compound (III) can be obtained when the phase transfer reagent, tetrabutylammonium hydroxide is employed as base.
Example II
N-Methyl-N-( 1-nanhthylmethvl)-2-hvdroxv-hentan-4-vnvl-1-amine f IIl To a solution of 3,3-dimethylbutyne (2.95 mL) in dry THF (50 mL) at -78°C
was added a 2.5 M solution of n-BuLi in hexane (10 mL) dropwise. The mixture was allowed to warm to room temperature over 15 min and stirred at that temperature for a further 15 min, then was cooled back to -78°C and BF3.OEt2 (3 mL) was added dropwise. The mixture was stirred for 15 min and 1.8 g of N-Methyl-N-naphthylmethyl-2,3-epoxypropane (III), dissolved in THF (10 mL), was added dropwise. After stirring at -78°C for 2 h, saturated sodium bicarbonate solution (1 S mL) was added, and the reaction mixture was allowed to warm to room temperature. The mixture was extracted with ethyl acetate (2x25 mL), and the combined organic fractions was dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatrography on silica gel (grade 9385, Merck, 230-400 mesh, 60 ~) using a mixture of hexane and ethyl acetate (85:15) as eluent, thereby affording the title compound as an oil (1.95 g, 79%).
'H-NMR (CDC13) 8 (ppm): 8.23 (m, 1H); 7.78-7.87 (m, 2H); 7.40-7.57 (m, 4H);
4.08 (d, J=13.0 Hz, 1 H); 3.92 (d, J=13 Hz, 1 H); 3.82 (m, 1 H); 3.62 (m, 1 H);
3.48 (m, 1 H);
2.60 (d, J=6.5 Hz, 2H); 2.31 (s, 3H) and 1.22 (s, 9H).
'3C-NMR (CDC13) 8 (ppm): 134.2, 134.0, 132.4, 128.6, 128.3, 127.7, 126.1, 125.7, 125.1, 124.3, 91.2, 74.2, 66.4, 62.4, 61.4, 42.3, 31.3, 27.4, 22.7.

HRMS: calc. for C,,H,,NO 309.2093 found 309.2108 Example III
N-methyl-N-(1-naphthylmethyl)-6,6-dimethylhept-2-en-4-ynyl-1-amine (I) (i) To an ice-cooled solution ofN-methyl-N-( 1-naphthylmethyl)-2-hydroxy-heptan-4-ynyl-1-amine (II) (155 mg) in THF (10 mL) was added Et3N (0.35 mL) followed by methanesulfonyl chloride (0.075 mL). The resulting mixture was stirred at 0°C for 3 h, then filtered. The filtrate was concentrated in vacuo, dissolved in toluene ( 10 mL) and DBU (0.37 mL) was added. The resulting mixture was heated at 80°C for 4 h, cooled to room temperature then poured onto a silica gel column and eluted with hexane ( 100%) followed by a mixture of hexane and ethyl acetate (95:5). Thus, a mixture of E- and Z- isomers o f N-methyl-N-( 1-naphthylmethyl)-6,6-dimethylhept-2-en-4-ynyl)-1-amine were obtained in a ratio of 2:5 (95 mg, 66%).
E- isomer (Ia); N-Methyl-N-( 1-naphthylmethyl)-6,6-dimethyl-hept-2(trans)-en-4-ynyl)-1-amine 'H-NMR (CDC13) 8 (ppm): 8.30 (m, 1 H); 7.80-7.86 (m, 2H); 7.27-7.61 (m, 4H);
6.20-6.29 (dt, J=16.0 and 6.5 Hz, 1 H), 5.75 (dt, J=16.0 and 1.5 Hz, 1 H), 3.9 ~
(s, 2H); 3.20 (dd, J=6.5 and 1.5 Hz, 2H); 2.21 (s, 3H) and 1.33 (s, 9H).
'3C-NMR (CDC13) 8 (ppm): 134.9, 134.8, 133.9, 132.5, 128.5, 128.0, 127.3, 125.9, 125.6, 125.2, 124.7, 112.9, 98.4, 77.3, 60.1, 59.7, 42.4, 31.1 and 28Ø
Z- isomer (Ib); N-Methyl-N-( 1-naphthylmethyl)-6,6-dimethyl-hept-2(cis)-en-4-ynyl)-1-amine 'H-NMR (CDC13) 8 (ppm): 8.25 (m, 1H); 7.71-7.86 (m, 2H); 7.38-7.55 (m, 4H);
6.00 6.09 (dt, J=10.8 and 6.5 Hz, 1 H), 5.66 (dt, J=10.8 and 1.4 Hz, 1 H), 3.93 (s, 2H); 3.38 (dd, J=6.9 and 1.3 Hz, 2H); 2.26 (s, 3H) and 1.28 (s, 9H).
(ii) When an ice-cooled solution of N-methyl-N-(1-naphthylmethyl)-2-hydroxy-heptan-4-ynyl-1-amine (II) in DMSO and THF (3:1 v/v) is treated with p-toluenesulfonyl chloride and solid KOH, a 1:4 mixture of E/Z terbinafine is obtained.
(iii) N-Methyl-N-(1-naphthylmethyl)-2-hydroxy-heptan-4-ynyl-1-amine (II) can be converted to its chloride by reacting with thionyl chloride and pyridine preferably at 0°C. Reaction of the resulting chloride with DBU in DMSO at 100°C afforded a 1:1 mixture of E/Z terbinafine.
(iv) When a solution of N-methyl-N-( 1-naphthylmethyl)-2-hydroxy-heptan-4-ynyl-1-amine (II) in DMSO is heated at 100°C for 16 h, a 1:9 mixture of E/Z
terbinafine is obtained.
(v) When a solution of N-methyl-N-( 1-naphthylmethyl)-2-hydroxy-heptan-4-ynyl-1-amine (II) in DMSO containing silica gel and p-toluenesulfonic acid is heated at 100°C for 16 h, a 1:1 mixture of E/Z terbinafine is obtained.
(vi) When a solution of N-methyl-N-(1-naphthylmethyl)-2-hydroxy-heptan-4-ynyl-1-amine (II) in toluene containing Amberlyst 15 is heated at 100°C for 36 h, a mixture of E/Z terbinafine is obtained.
Example IV
2-IN-methyl-N-naphth ImethYl]-dimethox et~~IV~
This reaction can be carried out according to the procedure described in Sandier and Karo. In an ice-cooled flask containing N-methyl-1-naphthalenemethylamine hydrochloride in anhydrous toluene is added 2 equivalents of sodium hydroxide followed by dropwise addition of bromoacetaldehyde dimethoxy acetal. The reaction mixture is then heated in a water bath at 80°C for 4 h then is cooled to room temperature. The reaction mixture is washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo. Purification of the residue by column chromatography on silica gel with a 10% EtOAc and hexane affords the title compound (IV).
In a similar manner, the reaction ofN-methyl-1-naphthalenemethylamine hydrochloride with bromoacetaldehyde ethylene acetal gives methylnaphthylmethyl-[ 1,3 ]-dioxolan-2-ylmethylamine.
Example V
2-fN-methyl-N-naphth Imethyll-acetaldehyde,~Vl (i) This reaction can be carried out according to the procedure described in Sandler and Karo. In an ice-cooled flask containing N-methyl-1-naphthalenemethylamine hydrochloride in anhydrous toluene is added 2 equivalents of sodium hydroxide followed by dropwise addition of bromoacetaldehyde dimethoxy acetal. The reaction mixture is then heated in a water bath at 80°C for 4 h then is cooled to room temperature. A 1:1 mixture of concentrated HCl and water is added and the reaction mixture is stirred at room temperature for 16 h. The aqueous layer is separated and the toluene layer is extracted twice with 10% hydrochloric acid. The combined aqueous layers is cooled and made alkaline with 40% sodium hydroxide solution, then extracted with ethyl acetate. The organic layer is collected, dried over sodium sulfate, filtered and concentrated in vacuo. Purification by column chromatography on silica gel using 10%
EtOAc and hexane affords the title compound (V).
Similar results are obtained when bromoacetaldehyde ethylene acetal is used as the alkylating reagent.
(ii) The procedure taken from Greene, is used. A mixture of 2-[N-methyl-N-naphthylmethyl]-dimethoxyethane in acetone and catalytic amount of p-toluenesulfonic acid is stirred at room temperature for 24 h. The reaction mixture is made alkaline with triethylamine and volatile materials are removed in vacuo. The residue is purified by column chromatography on silica gel using 10% EtOAc and hexane to give the title compound (V).
Example VI
N-Methvl-N-(1-naohthylmeth~l)-6 6-(dimethYl heft 2 en 4 yn~) 1 amine (i) This reaction is carried out according to the procedure of Corey et. al. n-BuLi is added to a solution of 4,4-dimethylpent-2-ynyl phosphonium bromide in dry THF at -20°C. The mixture is stirred under nitrogen for 30 min, and a solution of 2-[N-methyl-N-naphthylmethyl]-acetaldehyde in THF is added. The resulting mixture is stirred at room temperature for 5 h, then quenched by addition of saturated ammonium chloride 3 0 solution and extracted with ethyl acetate. The organic layer is dried over sodium sulfate - IS -and evaporated to give an oil. Column chromatography (5% EtOAc; hexane) gives the title compound as E/Z mixture.
(ii) In a similar fashion, an E/Z mixture of the title compound can be obtained by reacting a solution of 2-[N-methyl-N-naphthylmethyl]-acetaldehyde in THF with the reagent (VII).

Claims (18)

  1. THE EMBODIMENT OF THE PRESENT INVENTION IN WHICH
    EXCLUSIVE PROPERTIES AND RIGHT ARE CLAIMED ARE DEFINED
    AS FOLLOWS:

    A process for the manufacture of compounds of formula I:
    wherein R1 is lower alkyl, R2 is alkyl, arylalkyl, which comprises the following steps:
    (a) reacting N-alkyl-N-naphthylmethylamine with epichlorohydrin to give a compound of formula (III) in presence of a strong base and alcohol;
    wherein R1 is as defined above, and (b) reacting the compound of formula (III) with lithium alkylacetylide in the presence of lewis acid to give a compound of formula (II);
    wherein R1 and R2 are as defined above, then, (c) dehydrating compound of formula (II) into a compound of formula (I).
  2. 2. A process of claim 1 wherein the base is selected from the group consisting of sodium hydroxide, sodium methoxide and tetrabutylammonium hydroxide.
  3. 3. A process of claim 2 wherein the alcohol is a methanol.
  4. 4. A process of claim 3 wherein the lewis acid is boron trifluoride etherate.
  5. 5. A process of claim 4 wherein R1 is methyl.
  6. 6. A process of claim 5 wherein R2 is 1,1-dimethylethyl.
  7. 7. N-methyl-N(1-naphtylmethyl)-6,6 dimethyl-2-hydroxy-heptan-4-ynyl 1-amine.
  8. 8. N-lower alkyl-N(1 naphthylmethyl)-2-hydroxyalkyl-4-ynyl 1-amine.
  9. 9. A process for the manufacture of terbinafine which comprises the following steps:
    a. reacting N-methyl-N-naphthylmethylamine with epichlorohydrin to give N-methyl-N-naphthylmethyl-2,3-epoxypropane (III) in the presence of a strong base and an alcohol;
    b. reacting N-methyl-N-naphthylmethyl-2,3-epoxypropane (III) with lithium tert-butylacetylene to give N-Methyl-N-(1-naphthylmethyl)-6,6-dimethyl-2-hydroxyheptan-4-ynyl-1-amine (II);
    c. converting N-methyl-N-(1-naphthylmethyl)-6,6-dimethyl-2-hydroxyheptan-4-ynyl-1-amine (II) to terbinafme.
  10. 10. The process of claim 9, wherein the base is selected from the group consisting of sodium hydroxide, sodium methoxide and tetrabutylammonium hydroxide.
  11. 11. A process of claim 9, wherein the temperature of the reaction is 60°C.
  12. 12. A process of claim 9, wherein the alcohol is methanol.
  13. 13. A process of claim 9, wherein the reaction b is done in the presence of boron trifluoride etherate.
  14. 14. A process of claim 9, wherein the conversions of N-methyl-N-(1-naphthyl-methyl)-6,6-dimethyl-2-hydroxyheptan-4-ynyl-1-amine to terbinafine is made with methanesulfonyl chloride or toluenesulfonyl chloride in the presence of base.
  15. 15. A process of claim 14, wherein the base is selected from the group consisting of triethylamine, DBU, tent-butoxide and potassium hydroxide.
  16. 16. A process according to claim 9, wherein the conversion of N-methyl-N-(1-naphthylmethyl)-6,6-dimethyl-2-hydroxyheptan-4-ynyl-1-amine to terbinafine is a dehydration obtained either:
    (i) by heating N-methyl-N-(1-naphthylmethyl)-6,6-dimethyl-2-hydroxyheptan-4-ynyl-1-amine (II) with an acid in an organic solvent;
    or (ii) by heating N-methyl-N-(1-naphthylmethyl)-6,6-dimethyl-2-hydroxyheptan-4-ynyl-1-amine in dimethyl sulfoxide (DMSO).
  17. 17. The process according to claim 16, wherein the organic solvent is dimethyl sulfoxide or toluene.
  18. 18. The process according to claim 16 wherein heating is done at 100°C.
CA002185599A 1995-09-20 1996-09-16 Methods for the manufacture of terbinafine, trans-n-methyl-n-(1-naphthylmethyl)-6,6-dimethylhept-2-en-4 -ynyl-1-amine and pharmaceutically acceptable salt thereof, intermediates useful in the manufacture thereof, novel n-methyl-n-(1-naphthylmethyl)-6,6-dimethyl-2-hydroxyheptan- 4-ynyl-1-amine as potential antimycotic agents Expired - Fee Related CA2185599C (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
NZ280065 1995-09-20
NZ280065A NZ280065A (en) 1995-09-20 1995-09-20 Preparation of n-alkyl-n-(1-naphthylmethyl)alk-2-en-4-ynylamine derivatives

Publications (2)

Publication Number Publication Date
CA2185599A1 CA2185599A1 (en) 1997-03-21
CA2185599C true CA2185599C (en) 1999-11-30

Family

ID=19925473

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002185599A Expired - Fee Related CA2185599C (en) 1995-09-20 1996-09-16 Methods for the manufacture of terbinafine, trans-n-methyl-n-(1-naphthylmethyl)-6,6-dimethylhept-2-en-4 -ynyl-1-amine and pharmaceutically acceptable salt thereof, intermediates useful in the manufacture thereof, novel n-methyl-n-(1-naphthylmethyl)-6,6-dimethyl-2-hydroxyheptan- 4-ynyl-1-amine as potential antimycotic agents

Country Status (3)

Country Link
US (1) US5817875A (en)
CA (1) CA2185599C (en)
NZ (1) NZ280065A (en)

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0003360D0 (en) * 2000-02-14 2000-04-05 Novartis Ag Monoclonal antibodies
SK5202000A3 (en) 2000-04-07 2001-12-03 Slovakofarma As Method for the preparation of (e)-n-(6,6-dimethyl-2-hepten-4- inyl)-n-methyl-1-naphthalenemethylamine (terbinaphin)
GB0103046D0 (en) * 2001-02-07 2001-03-21 Novartis Ag Organic Compounds
ITMI20010430A1 (en) * 2001-03-02 2002-09-02 Dinamite Dipharma S P A In For METHOD FOR THE SYNTHESIS OF TERBINAFINA
US7244703B2 (en) 2001-06-22 2007-07-17 Bentley Pharmaceuticals, Inc. Pharmaceutical compositions and methods for peptide treatment
AR034813A1 (en) * 2001-07-20 2004-03-17 Novartis Ag PHARMACEUTICAL COMPOSITIONS AND USE OF THE SAME
KR100459275B1 (en) 2001-12-28 2004-12-03 주식회사유한양행 A PROCESS FOR THE PREPARATION OF TERBINAFINE OR HCl SALT THEREOF
KR20030070768A (en) * 2002-02-26 2003-09-02 (주)리드젠 PREPARATION OF N-(4-t-BUTYLBENZYL)-N-METHYL-1-NAPHTHYLMETHYLAMINE HYDROCHLORIDE AND PURIFICATION THEREOF
GB0320312D0 (en) 2003-08-29 2003-10-01 Novartis Ag Purification process
US20050197512A1 (en) * 2003-08-29 2005-09-08 Ulrich Beutler Purification process
NZ547587A (en) 2003-12-08 2009-09-25 Cpex Pharmaceuticals Inc Pharmaceutical compositions and methods for insulin treatment
WO2005058794A1 (en) * 2003-12-17 2005-06-30 Natco Pharma Limited An improved process for the preparation of terbinafine intermediate
ITMI20041154A1 (en) * 2004-06-09 2004-09-09 Italiana Sint Spa SUMMARY PROCEDURE OF TERBINAFINA AND ITS DERIVATIVES
US20060004230A1 (en) * 2004-06-30 2006-01-05 Joseph Kaspi Process for the preparation of terbinafine and salts thereof
US20060078580A1 (en) 2004-10-08 2006-04-13 Mediquest Therapeutics, Inc. Organo-gel formulations for therapeutic applications
US7740875B2 (en) * 2004-10-08 2010-06-22 Mediquest Therapeutics, Inc. Organo-gel formulations for therapeutic applications
WO2012033956A1 (en) 2010-09-08 2012-03-15 Mithridion, Inc. Cognition enhancing compounds and compositions, methods of making, and methods of treating

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE19975055I1 (en) * 1979-08-22 2000-01-27 Novartis Ag Propenylamines Process for their preparation pharmaceutical compositions containing them and their use as medicines

Also Published As

Publication number Publication date
NZ280065A (en) 1998-04-27
CA2185599A1 (en) 1997-03-21
US5817875A (en) 1998-10-06

Similar Documents

Publication Publication Date Title
CA2185599C (en) Methods for the manufacture of terbinafine, trans-n-methyl-n-(1-naphthylmethyl)-6,6-dimethylhept-2-en-4 -ynyl-1-amine and pharmaceutically acceptable salt thereof, intermediates useful in the manufacture thereof, novel n-methyl-n-(1-naphthylmethyl)-6,6-dimethyl-2-hydroxyheptan- 4-ynyl-1-amine as potential antimycotic agents
ES2378507T3 (en) Synthetic routes for 2 (S), 4 (S), 5 (S), 7 (S) -2,7-dialkyl-4-hydroxy-5-amino-8-aryl-octanoylamides
WO2008069440A1 (en) The preparation method of 2-de0xy-l-rib0se
US20030176710A1 (en) C1-C6-epothilone fragments and process for the production of C1-C6-fragments of epothilones and derivatives thereof
US20130310577A1 (en) Convergent synthesis of renin inhibitors and intermediates useful therein
KR980700956A (en) Method for preparing 2-(2-hydroxymethylphenyl)acetamide derivative and intermediate for the preparation (PROCESS FOR PRODUCING 2-(2-HYDROXYMETHYLPHENYL)ACETAMIDE DERIVATIVE AND INT ERMEDIATE FOR THE PRODUCTION THEREOF)
CN114315609B (en) Technological method for preparing cis-2-aminocyclohexanol
CN114560795B (en) Method for preparing cyclosulfamide
JPH11349541A (en) New intermediate for allosamizoline derivative
KR101130717B1 (en) Process for the Preparation of a Chiral Intermediate for the Preparation of HMG-CoA Reductase Inhibitors
RU2654054C1 (en) Method for production of 3-alkyl-3-azabicyclo[3,3,1]none-1(9),5,7-trien-9-ols or 3-alkyl-3,4-dihydro-2h-1,3-benzoxazines
Kamimura et al. syn-and anti-Selective preparation of 3-substltuted-Δ2-isoxazolines
EP2203434B1 (en) Method of preparing (6r)-3-hexyl-4-hydroxy-6-undecyl-5,6-dihydropyran-2-one, and intermediate used in the method
AU2005209382B2 (en) Method for producing a 2-(ethoxymethyl)tropane derivative
CA2463232A1 (en) Process for producing (2-nitro-phenyl)acetonirtile derivative and intermediate used for synthesis thereof
EP3956332B1 (en) Diasteroselective process for the preparation of thiol- or disulfide-containing maytansinoid esters and intermediates thereof
CN114213323B (en) New process for synthesizing procaterol hydrochloride
KR100203457B1 (en) Process for terbinafine
KR100763770B1 (en) Process for preparing chiral intermediates useful in synthesis of atorvastatin
KR100856133B1 (en) Improved process for preparing atorvastatin
CN117342978A (en) Cyanomethylation method of amine
JP2000063321A (en) Production of long-chain beta-hydroxycarboxylic acid of high optical purity
US6281379B1 (en) Process for producing norstatin derivatives
CN116924997A (en) Preparation method of azoxystrobin intermediate
CN116891461A (en) Synthesis method of SERD intermediate

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed